1
|
Ayoub S, Arabi M, Al-Najjar Y, Laswi I, Outeiro TF, Chaari A. Glycation in Alzheimer's Disease and Type 2 Diabetes: The Prospect of Dual Drug Approaches for Therapeutic Interventions. Mol Neurobiol 2025:10.1007/s12035-025-05051-9. [PMID: 40402411 DOI: 10.1007/s12035-025-05051-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
As global life expectancy increases, the prevalence of neurodegenerative diseases like Alzheimer's disease (AD) continues to rise. Since therapeutic options are minimal, a deeper understanding of the pathophysiology is essential for improved diagnosis and treatments. AD is marked by the aggregation of Aβ proteins, tau hyperphosphorylation, and progressive neuronal loss, though its precise origins remain poorly understood. Meanwhile, type 2 diabetes mellitus (T2DM) is characterized by chronic hyperglycemia, leading to the formation of advanced glycation end products (AGEs), which are implicated in tissue damage and neurotoxicity. These AGEs can be resistant to proteolysis and, therefore, accumulate, exacerbating AD pathology and accelerating neurodegeneration. Insulin resistance, a hallmark of T2DM, further complicates AD pathogenesis by promoting tau hyperphosphorylation and Aβ plaque accumulation. Additionally, gut microbiome dysbiosis in T2DM fosters AGE accumulation and neuroinflammation, underscoring the intricate relationship between metabolic disorders, gut health, and neurodegenerative processes. This complex interplay presents both a challenge and a potential avenue for therapeutic intervention. Emerging evidence suggests that antidiabetic medications may offer cognitive benefits in AD, as well as in other neurodegenerative conditions, pointing to a shared pathophysiology. Thus, we posit that targeting AGEs, insulin signaling, and gut microbiota dynamics presents promising opportunities for innovative treatment approaches in AD and T2DM.
Collapse
Affiliation(s)
- Sama Ayoub
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Maryam Arabi
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Yousef Al-Najjar
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Ibrahim Laswi
- Department of Internal Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, Newcastle, NE2 4HH, UK
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Scientific Employee With an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Straße 3a, 37075, Göttingen, Germany
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|
2
|
Kciuk M, Kruczkowska W, Gałęziewska J, Wanke K, Kałuzińska-Kołat Ż, Aleksandrowicz M, Kontek R. Alzheimer's Disease as Type 3 Diabetes: Understanding the Link and Implications. Int J Mol Sci 2024; 25:11955. [PMID: 39596023 PMCID: PMC11593477 DOI: 10.3390/ijms252211955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two prevalent conditions that present considerable public health issue in aging populations worldwide. Recent research has proposed a novel conceptualization of AD as "type 3 diabetes", highlighting the critical roles of insulin resistance and impaired glucose metabolism in the pathogenesis of the disease. This article examines the implications of this association, exploring potential new avenues for treatment and preventive strategies for AD. Key evidence linking diabetes to AD emphasizes critical metabolic processes that contribute to neurodegeneration, including inflammation, oxidative stress, and alterations in insulin signaling pathways. By framing AD within this metabolic context, we can enhance our understanding of its etiology, which in turn may influence early diagnosis, treatment plans, and preventive measures. Understanding AD as a manifestation of diabetes opens up the possibility of employing novel therapeutic strategies that incorporate lifestyle modifications and the use of antidiabetic medications to mitigate cognitive decline. This integrated approach has the potential to improve patient outcomes and deepen our comprehension of the intricate relationship between neurodegenerative diseases and metabolic disorders.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Weronika Kruczkowska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Julia Gałęziewska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Katarzyna Wanke
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, 90-136 Lodz, Poland
| | - Marta Aleksandrowicz
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| |
Collapse
|
3
|
Moore A, Ritchie MD. Cross-phenotype associations between Alzheimer's Disease and its comorbidities may provide clues to progression. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2024; 2024:623-631. [PMID: 38827078 PMCID: PMC11141840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, with one in nine people over the age of 65 living with the disease in 2023. In this study, we used a phenome wide association study (PheWAS) approach to identify cross-phenotype between previously identified genetic associations for AD and electronic health record (EHR) diagnoses from the UK Biobank (UKBB) (n=361,194 of European ancestry) and the eMERGE Network (n=105,108 of diverse ancestry). Based on 497 previously identified AD-associated variants from the Alzheimer's Disease Variant Portal (ADVP), we found significant associations primarily in immune and cardiac related diseases in our PheWAS. Replicating variants have widespread impacts on immune genes in diverse tissue types. This study demonstrates the potential of using the PheWAS strategy to improve our understanding of AD progression as well as identify potential drug repurposing opportunities for new treatment and disease prevention strategies.
Collapse
Affiliation(s)
- Anni Moore
- Genomics and Computational Biology Group, University of Pennsylvania, Philadelphia, PA
| | - Marylyn D Ritchie
- Genomics and Computational Biology Group, University of Pennsylvania, Philadelphia, PA
- Institute of Biomedical Informatics, University of Pennsylvania, Philadelphia, PA
- Division of Informatics, DBEI, Perelman School of Medicine., University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
4
|
Albar NY, Hassaballa H, Shikh H, Albar Y, Ibrahim AS, Mousa AH, Alshanberi AM, Elgebaly A, Bahbah EI. The interaction between insulin resistance and Alzheimer's disease: a review article. Postgrad Med 2024; 136:377-395. [PMID: 38804907 DOI: 10.1080/00325481.2024.2360887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Insulin serves multiple functions as a growth-promoting hormone in peripheral tissues. It manages glucose metabolism by promoting glucose uptake into cells and curbing the production of glucose in the liver. Beyond this, insulin fosters cell growth, drives differentiation, aids protein synthesis, and deters degradative processes like glycolysis, lipolysis, and proteolysis. Receptors for insulin and insulin-like growth factor-1 are widely expressed in the central nervous system. Their widespread presence in the brain underscores the varied and critical functions of insulin signaling there. Insulin aids in bolstering cognition, promoting neuron extension, adjusting the release and absorption of catecholamines, and controlling the expression and positioning of gamma-aminobutyric acid (GABA). Importantly, insulin can effortlessly traverse the blood-brain barrier. Furthermore, insulin resistance (IR)-induced alterations in insulin signaling might hasten brain aging, impacting its plasticity and potentially leading to neurodegeneration. Two primary pathways are responsible for insulin signal transmission: the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, which oversees metabolic responses, and the mitogen-activated protein kinase (MAPK) pathway, which guides cell growth, survival, and gene transcription. This review aimed to explore the potential shared metabolic traits between Alzheimer's disease (AD) and IR disorders. It delves into the relationship between AD and IR disorders, their overlapping genetic markers, and shared metabolic indicators. Additionally, it addresses existing therapeutic interventions targeting these intersecting pathways.
Collapse
Affiliation(s)
- Nezar Y Albar
- Internal Medicine Department, Dr. Samir Abbas Hospital, Jeddah, Saudi Arabia
| | | | - Hamza Shikh
- Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Yassin Albar
- Fakeeh College of Medical Sciences, Jeddah, Saudi Arabia
| | | | - Ahmed Hafez Mousa
- Department of Neurosurgery, Postgraduate Medical Education, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Department of Neurosurgery, Rashid Hospital, Dubai Academic Health Cooperation, Dubai, United Arab Emirates
| | - Asim Muhammed Alshanberi
- Department of Community Medicine and Pilgrims Health Care, Umm Alqura University, Makkah, Saudi Arabia
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ahmed Elgebaly
- Smart Health Academic Unit, University of East London, London, UK
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
5
|
Zhang X, Zhu Z, Huang Y, Shang X, O'Brien TJ, Kwan P, Ha J, Wang W, Liu S, Zhang X, Kiburg K, Bao Y, Wang J, Yu H, He M, Zhang L. Shared genetic aetiology of Alzheimer's disease and age-related macular degeneration by APOC1 and APOE genes. BMJ Neurol Open 2024; 6:e000570. [PMID: 38646507 PMCID: PMC11029327 DOI: 10.1136/bmjno-2023-000570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/04/2024] [Indexed: 04/23/2024] Open
Abstract
Background Alzheimer's disease (AD) and age-related macular degeneration (AMD) share similar pathological features, suggesting common genetic aetiologies between the two. Investigating gene associations between AD and AMD may provide useful insights into the underlying pathogenesis and inform integrated prevention and treatment for both diseases. Methods A stratified quantile-quantile (QQ) plot was constructed to detect the pleiotropy among AD and AMD based on genome-wide association studies data from 17 008 patients with AD and 30 178 patients with AMD. A Bayesian conditional false discovery rate-based (cFDR) method was used to identify pleiotropic genes. UK Biobank was used to verify the pleiotropy analysis. Biological network and enrichment analysis were conducted to explain the biological reason for pleiotropy phenomena. A diagnostic test based on gene expression data was used to predict biomarkers for AD and AMD based on pleiotropic genes and their regulators. Results Significant pleiotropy was found between AD and AMD (significant leftward shift on QQ plots). APOC1 and APOE were identified as pleiotropic genes for AD-AMD (cFDR <0.01). Network analysis revealed that APOC1 and APOE occupied borderline positions on the gene co-expression networks. Both APOC1 and APOE genes were enriched on the herpes simplex virus 1 infection pathway. Further, machine learning-based diagnostic tests identified that APOC1, APOE (areas under the curve (AUCs) >0.65) and their upstream regulators, especially ZNF131, ADNP2 and HINFP, could be potential biomarkers for both AD and AMD (AUCs >0.8). Conclusion In this study, we confirmed the genetic pleiotropy between AD and AMD and identified APOC1 and APOE as pleiotropic genes. Further, the integration of multiomics data identified ZNF131, ADNP2 and HINFP as novel diagnostic biomarkers for AD and AMD.
Collapse
Affiliation(s)
- Xueli Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Patrick Kwan
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jason Ha
- Alfred Health, Melbourne, Victoria, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Shunming Liu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Katerina Kiburg
- Centre for Eye Research, University of Melbourne, East Melbourne, Victoria, Australia
| | - Yining Bao
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing Wang
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Lei Zhang
- Clinical Medical Research Center, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Chen A, Li Q, Huang Y, Li Y, Chuang YN, Hu X, Guo S, Wu Y, Guo Y, Bian J. Feasibility of Identifying Factors Related to Alzheimer's Disease and Related Dementia in Real-World Data. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.10.24302621. [PMID: 38405723 PMCID: PMC10889002 DOI: 10.1101/2024.02.10.24302621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
A comprehensive view of factors associated with AD/ADRD will significantly aid in studies to develop new treatments for AD/ADRD and identify high-risk populations and patients for prevention efforts. In our study, we summarized the risk factors for AD/ADRD by reviewing existing meta-analyses and review articles on risk and preventive factors for AD/ADRD. In total, we extracted 477 risk factors in 10 categories from 537 studies. We constructed an interactive knowledge map to disseminate our study results. Most of the risk factors are accessible from structured Electronic Health Records (EHRs), and clinical narratives show promise as information sources. However, evaluating genomic risk factors using RWD remains a challenge, as genetic testing for AD/ADRD is still not a common practice and is poorly documented in both structured and unstructured EHRs. Considering the constantly evolving research on AD/ADRD risk factors, literature mining via NLP methods offers a solution to automatically update our knowledge map.
Collapse
Affiliation(s)
- Aokun Chen
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Qian Li
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yu Huang
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yongqiu Li
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yu-neng Chuang
- Department of Computer Science, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005
| | - Xia Hu
- Department of Computer Science, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005
| | - Serena Guo
- Department of Pharmaceutical Outcomes & Policy, College of Pharmacy, University of Florida, 1225 Center Drive, Gainesville, FL 32610
| | - Yonghui Wu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yi Guo
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| |
Collapse
|
7
|
Kuate Defo A, Bakula V, Pisaturo A, Labos C, Wing SS, Daskalopoulou SS. Diabetes, antidiabetic medications and risk of dementia: A systematic umbrella review and meta-analysis. Diabetes Obes Metab 2024; 26:441-462. [PMID: 37869901 DOI: 10.1111/dom.15331] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/12/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
AIMS The objective of this umbrella review and meta-analysis was to evaluate the effect of diabetes on risk of dementia, as well as the mitigating effect of antidiabetic treatments. MATERIALS AND METHODS We conducted a systematic umbrella review on diabetes and its treatment, and a meta-analysis focusing on treatment. We searched MEDLINE/PubMed, Embase, PsycINFO, CINAHL and the Cochrane Library for systematic reviews and meta-analyses assessing the risk of cognitive decline/dementia in individuals with diabetes until 2 July 2023. We conducted random-effects meta-analyses to obtain risk ratios and 95% confidence intervals estimating the association of metformin, thiazolidinediones, pioglitazone, dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors, meglitinides, insulin, sulphonylureas, glucagon-like peptide-1 receptor agonists (GLP1RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2is) with risk of dementia from cohort/case-control studies. The subgroups analysed included country and world region. Risk of bias was assessed with the AMSTAR tool and Newcastle-Ottawa Scale. RESULTS We included 100 reviews and 27 cohort/case-control studies (N = 3 046 661). Metformin, thiazolidinediones, pioglitazone, GLP1RAs and SGLT2is were associated with significant reduction in risk of dementia. When studies examining metformin were divided by country, the only significant effect was for the United States. Moreover, the effect of metformin was significant in Western but not Eastern populations. No significant effect was observed for dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors, or insulin, while meglitinides and sulphonylureas were associated with increased risk. CONCLUSIONS Metformin, thiazolidinediones, pioglitazone, GLP1RAs and SGLT2is were associated with reduced risk of dementia. More longitudinal studies aimed at determining their relative benefit in different populations should be conducted.
Collapse
Affiliation(s)
- Alvin Kuate Defo
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Veselko Bakula
- Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | - Christopher Labos
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Simon S Wing
- Division of Endocrinology & Metabolism, Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Stella S Daskalopoulou
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Division of Internal Medicine, Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Moore A, Ritchie MD. Cross-phenotype associations between Alzheimer's Disease and its comorbidities may provide clues to progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.06.23297993. [PMID: 37986758 PMCID: PMC10659497 DOI: 10.1101/2023.11.06.23297993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, with one in nine people over the age of 65 living with the disease in 2023. In this study, we used a phenome wide association study (PheWAS) approach to identify cross-phenotype associations between previously identified genetic AD and for electronic health record (EHR) diagnoses from the UK Biobank (UKBB) (n=361,194 of European ancestry) and the eMERGE Network (n=105,108 of diverse ancestry). Based on 497 previously identified AD-associated variants from the Alzheimer's Disease Variant Portal (ADVP), we found significant associations primarily in immune and cardiac related diseases in our PheWAS. Replicating variants have widespread impacts on immune genes in diverse tissue types. This study demonstrates the potential of using the PheWAS strategy to improve our understanding of AD progression as well as identify potential drug repurposing opportunities for new treatment and disease prevention strategies.
Collapse
Affiliation(s)
- Anni Moore
- Genomics and Computational Biology Group, University of Pennsylvania, Philadelphia, PA
| | - Marylyn D Ritchie
- Genomics and Computational Biology Group, University of Pennsylvania, Philadelphia, PA
- Institute of Biomedical Informatics, University of Pennsylvania, Philadelphia, PA; Division of Informatics, DBEI, Perelman School of Medicine., University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
9
|
Lin D, Liu X, Li Q, Qin J, Xiong Z, Wu X. Association between gut microbiome and intracerebral hemorrhage based on genome-wide association study data. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1176-1184. [PMID: 37875357 PMCID: PMC10930854 DOI: 10.11817/j.issn.1672-7347.2023.230107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Indexed: 10/26/2023]
Abstract
OBJECTIVES Intracerebral hemorrhage (ICH) has the highest mortality and disability rates among various subtypes of stroke. Previous studies have shown that the gut microbiome (GM) is closely related to the risk factors and pathological basis of ICH. This study aims to explore the causal effect of GM on ICH and the potential mechanisms. METHODS Genome wide association study (GWAS) data on GM and ICH were obtained from Microbiome Genome and International Stroke Genetics Consortium. Based on the GWAS data, we first performed Mendelian randomization (MR) analysis to evaluate the causal association between GM and ICH. Then, a conditional false discovery rate (cFDR) method was conducted to identify the pleiotropic variants. RESULTS MR analysis showed that Pasteurellales, Pasteurellaceae, and Haemophilus were negatively correlated with the risk of ICH, whileVerrucomicrobiae, Verrucomicrobiales, Verrucomicrobiaceae, Akkermansia, Holdemanella, and LachnospiraceaeUCG010 were positively correlated with ICH. By applying the cFDR method, 3 pleiotropic loci (rs331083, rs4315115, and rs12553325) were found to be associated with both GM and ICH. CONCLUSIONS There is a causal association and pleiotropic variants between GM and ICH.
Collapse
Affiliation(s)
- Dihui Lin
- Department of Public Health and Medical Technology, College of Medicine, Jishou University, Jishou Hunan 416000.
| | - Xinpeng Liu
- Department of Public Health and Medical Technology, College of Medicine, Jishou University, Jishou Hunan 416000
| | - Qi Li
- Xiangxi Tujia and Miao Autonomous Prefecture Center for Disease Prevention and Control, Jishou Hunan 416000
| | - Jiabi Qin
- Department of Epidemic and Health Statistics, Xiangya School of Public Health, Central South University, Changsha 410006, China
| | - Zhendong Xiong
- Department of Public Health and Medical Technology, College of Medicine, Jishou University, Jishou Hunan 416000
| | - Xinrui Wu
- Department of Public Health and Medical Technology, College of Medicine, Jishou University, Jishou Hunan 416000.
| |
Collapse
|
10
|
Horimoto AR, Boyken LA, Blue EE, Grinde KE, Nafikov RA, Sohi HK, Nato AQ, Bis JC, Brusco LI, Morelli L, Ramirez A, Dalmasso MC, Temple S, Satizabal C, Browning SR, Seshadri S, Wijsman EM, Thornton TA. Admixture mapping implicates 13q33.3 as ancestry-of-origin locus for Alzheimer disease in Hispanic and Latino populations. HGG ADVANCES 2023; 4:100207. [PMID: 37333771 PMCID: PMC10276158 DOI: 10.1016/j.xhgg.2023.100207] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Alzheimer disease (AD) is the most common form of senile dementia, with high incidence late in life in many populations including Caribbean Hispanic (CH) populations. Such admixed populations, descended from more than one ancestral population, can present challenges for genetic studies, including limited sample sizes and unique analytical constraints. Therefore, CH populations and other admixed populations have not been well represented in studies of AD, and much of the genetic variation contributing to AD risk in these populations remains unknown. Here, we conduct genome-wide analysis of AD in multiplex CH families from the Alzheimer Disease Sequencing Project (ADSP). We developed, validated, and applied an implementation of a logistic mixed model for admixture mapping with binary traits that leverages genetic ancestry to identify ancestry-of-origin loci contributing to AD. We identified three loci on chromosome 13q33.3 associated with reduced risk of AD, where associations were driven by Native American (NAM) ancestry. This AD admixture mapping signal spans the FAM155A, ABHD13, TNFSF13B, LIG4, and MYO16 genes and was supported by evidence for association in an independent sample from the Alzheimer's Genetics in Argentina-Alzheimer Argentina consortium (AGA-ALZAR) study with considerable NAM ancestry. We also provide evidence of NAM haplotypes and key variants within 13q33.3 that segregate with AD in the ADSP whole-genome sequencing data. Interestingly, the widely used genome-wide association study approach failed to identify associations in this region. Our findings underscore the potential of leveraging genetic ancestry diversity in recently admixed populations to improve genetic mapping, in this case for AD-relevant loci.
Collapse
Affiliation(s)
| | - Lisa A. Boyken
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth E. Blue
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Kelsey E. Grinde
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Department of Mathematics, Statistics and Computer Science, Macalester College, Saint Paul, MN 55105, USA
| | - Rafael A. Nafikov
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Harkirat K. Sohi
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Biomedical and Health Informatics Program, University of Washington, Seattle, WA 98195, USA
| | - Alejandro Q. Nato
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Luis I. Brusco
- CENECON - Center of Behavioural Neurology and Neuropsychiatry, School of Medicine, University of Buenos Aires, C1121A6B Buenos Aires, Argentina
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration-Fundación Instituto Leloir-IIBBA- National Scientific and Technical Research Council (CONICET), C1405BWE Ciudad Autónoma de Buenos Aires, Argentina
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) University of Cologne, 50674 Cologne, Germany
- Department of Psychiatry, UT Health San Antonio, San Antonio, TX 78229, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Maria Carolina Dalmasso
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
- Neurosciences and Complex Systems Unit (EnyS), CONICET, Hospital El Cruce, National University A. Jauretche (UNAJ), B1888AAE Florencio Varela, Argentina
| | - Seth Temple
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - Claudia Satizabal
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas, San Antonio, TX 78229, USA
- Department of Neurology, University of Texas, San Antonio, TX 78229, USA
| | - Sharon R. Browning
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Sudha Seshadri
- Department of Neurology, University of Texas, San Antonio, TX 78229, USA
| | - Ellen M. Wijsman
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Fominykh V, Shadrin AA, Jaholkowski PP, Bahrami S, Athanasiu L, Wightman DP, Uffelmann E, Posthuma D, Selbæk G, Dale AM, Djurovic S, Frei O, Andreassen OA. Shared genetic loci between Alzheimer's disease and multiple sclerosis: Crossroads between neurodegeneration and immune system. Neurobiol Dis 2023; 183:106174. [PMID: 37286172 PMCID: PMC11884797 DOI: 10.1016/j.nbd.2023.106174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/27/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Neuroinflammation is involved in the pathophysiology of Alzheimer's disease (AD), including immune-linked genetic variants and molecular pathways, microglia and astrocytes. Multiple Sclerosis (MS) is a chronic, immune-mediated disease with genetic and environmental risk factors and neuropathological features. There are clinical and pathobiological similarities between AD and MS. Here, we investigated shared genetic susceptibility between AD and MS to identify putative pathological mechanisms shared between neurodegeneration and the immune system. METHODS We analysed GWAS data for late-onset AD (N cases = 64,549, N controls = 634,442) and MS (N cases = 14,802, N controls = 26,703). Gaussian causal mixture modelling (MiXeR) was applied to characterise the genetic architecture and overlap between AD and MS. Local genetic correlation was investigated with Local Analysis of [co]Variant Association (LAVA). The conjunctional false discovery rate (conjFDR) framework was used to identify the specific shared genetic loci, for which functional annotation was conducted with FUMA and Open Targets. RESULTS MiXeR analysis showed comparable polygenicities for AD and MS (approximately 1800 trait-influencing variants) and genetic overlap with 20% of shared trait-influencing variants despite negligible genetic correlation (rg = 0.03), suggesting mixed directions of genetic effects across shared variants. conjFDR analysis identified 16 shared genetic loci, with 8 having concordant direction of effects in AD and MS. Annotated genes in shared loci were enriched in molecular signalling pathways involved in inflammation and the structural organisation of neurons. CONCLUSIONS Despite low global genetic correlation, the current results provide evidence for polygenic overlap between AD and MS. The shared loci between AD and MS were enriched in pathways involved in inflammation and neurodegeneration, highlighting new opportunities for future investigation.
Collapse
Affiliation(s)
- Vera Fominykh
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Alexey A Shadrin
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Piotr P Jaholkowski
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Shahram Bahrami
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lavinia Athanasiu
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Douglas P Wightman
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Emil Uffelmann
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Geir Selbæk
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway; Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tonsberg, Vestfold, Norway
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, California, USA; Multimodal Imaging Laboratory, University of California San Diego, La Jolla, California, USA; Department of Psychiatry, University of California San Diego, La Jolla, California, USA; Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Srdjan Djurovic
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Oleksandr Frei
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Informatics, Centre for Bioinformatics, University of Oslo, Norway
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Boukhalfa W, Jmel H, Kheriji N, Gouiza I, Dallali H, Hechmi M, Kefi R. Decoding the genetic relationship between Alzheimer's disease and type 2 diabetes: potential risk variants and future direction for North Africa. Front Aging Neurosci 2023; 15:1114810. [PMID: 37342358 PMCID: PMC10277480 DOI: 10.3389/fnagi.2023.1114810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/11/2023] [Indexed: 06/22/2023] Open
Abstract
Introduction Alzheimer's disease (AD) and Type 2 diabetes (T2D) are both age-associated diseases. Identification of shared genes could help develop early diagnosis and preventive strategies. Although genetic background plays a crucial role in these diseases, we noticed an underrepresentation tendency of North African populations in omics studies. Materials and methods First, we conducted a comprehensive review of genes and pathways shared between T2D and AD through PubMed. Then, the function of the identified genes and variants was investigated using annotation tools including PolyPhen2, RegulomeDB, and miRdSNP. Pathways enrichment analyses were performed with g:Profiler and EnrichmentMap. Next, we analyzed variant distributions in 16 worldwide populations using PLINK2, R, and STRUCTURE software. Finally, we performed an inter-ethnic comparison based on the minor allele frequency of T2D-AD common variants. Results A total of 59 eligible papers were included in our study. We found 231 variants and 363 genes shared between T2D and AD. Variant annotation revealed six single nucleotide polymorphisms (SNP) with a high pathogenic score, three SNPs with regulatory effects on the brain, and six SNPs with potential effects on miRNA-binding sites. The miRNAs affected were implicated in T2D, insulin signaling pathways, and AD. Moreover, replicated genes were significantly enriched in pathways related to plasma protein binding, positive regulation of amyloid fibril deposition, microglia activation, and cholesterol metabolism. Multidimensional screening performed based on the 363 shared genes showed that main North African populations are clustered together and are divergent from other worldwide populations. Interestingly, our results showed that 49 SNP associated with T2D and AD were present in North African populations. Among them, 11 variants located in DNM3, CFH, PPARG, ROHA, AGER, CLU, BDNF1, CST9, and PLCG1 genes display significant differences in risk allele frequencies between North African and other populations. Conclusion Our study highlighted the complexity and the unique molecular architecture of North African populations regarding T2D-AD shared genes. In conclusion, we emphasize the importance of T2D-AD shared genes and ethnicity-specific investigation studies for a better understanding of the link behind these diseases and to develop accurate diagnoses using personalized genetic biomarkers.
Collapse
Affiliation(s)
- Wided Boukhalfa
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Haifa Jmel
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
| | - Nadia Kheriji
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Ismail Gouiza
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis, Tunisia
- University of Angers, MitoLab Team, Unité MitoVasc, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France
| | - Hamza Dallali
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
| | - Mariem Hechmi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
| | - Rym Kefi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
13
|
Amin AM, Mostafa H, Khojah HMJ. Insulin resistance in Alzheimer's disease: The genetics and metabolomics links. Clin Chim Acta 2023; 539:215-236. [PMID: 36566957 DOI: 10.1016/j.cca.2022.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with significant socioeconomic burden worldwide. Although genetics and environmental factors play a role, AD is highly associated with insulin resistance (IR) disorders such as metabolic syndrome (MS), obesity, and type two diabetes mellitus (T2DM). These findings highlight a shared pathogenesis. The use of metabolomics as a downstream systems' biology (omics) approach can help to identify these shared metabolic traits and assist in the early identification of at-risk groups and potentially guide therapy. Targeting the shared AD-IR metabolic trait with lifestyle interventions and pharmacological treatments may offer promising AD therapeutic approach. In this narrative review, we reviewed the literature on the AD-IR pathogenic link, the shared genetics and metabolomics biomarkers between AD and IR disorders, as well as the lifestyle interventions and pharmacological treatments which target this pathogenic link.
Collapse
Affiliation(s)
- Arwa M Amin
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah, Saudi Arabia.
| | - Hamza Mostafa
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Hani M J Khojah
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
14
|
Ye XW, Liu MN, Wang X, Cheng SQ, Li CS, Bai YY, Yang LL, Wang XX, Wen J, Xu WJ, Zhang SY, Xu XF, Li XR. Exploring the common pathogenesis of Alzheimer's disease and type 2 diabetes mellitus via microarray data analysis. Front Aging Neurosci 2023; 15:1071391. [PMID: 36923118 PMCID: PMC10008874 DOI: 10.3389/fnagi.2023.1071391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
Background Alzheimer's Disease (AD) and Type 2 Diabetes Mellitus (DM) have an increased incidence in modern society. Although more and more evidence has supported that DM is prone to AD, the interrelational mechanisms remain fully elucidated. Purpose The primary purpose of this study is to explore the shared pathophysiological mechanisms of AD and DM. Methods Download the expression matrix of AD and DM from the Gene Expression Omnibus (GEO) database with sequence numbers GSE97760 and GSE95849, respectively. The common differentially expressed genes (DEGs) were identified by limma package analysis. Then we analyzed the six kinds of module analysis: gene functional annotation, protein-protein interaction (PPI) network, potential drug screening, immune cell infiltration, hub genes identification and validation, and prediction of transcription factors (TFs). Results The subsequent analyses included 339 common DEGs, and the importance of immunity, hormone, cytokines, neurotransmitters, and insulin in these diseases was underscored by functional analysis. In addition, serotonergic synapse, ovarian steroidogenesis, estrogen signaling pathway, and regulation of lipolysis are closely related to both. DEGs were input into the CMap database to screen small molecule compounds with the potential to reverse AD and DM pathological functions. L-690488, exemestane, and BMS-345541 ranked top three among the screened small molecule compounds. Finally, 10 essential hub genes were identified using cytoHubba, including PTGS2, RAB10, LRRK2, SOS1, EEA1, NF1, RAB14, ADCY5, RAPGEF3, and PRKACG. For the characteristic Aβ and Tau pathology of AD, RAPGEF3 was associated significantly positively with AD and NF1 significantly negatively with AD. In addition, we also found ADCY5 and NF1 significant correlations with DM phenotypes. Other datasets verified that NF1, RAB14, ADCY5, and RAPGEF3 could be used as key markers of DM complicated with AD. Meanwhile, the immune cell infiltration score reflects the different cellular immune microenvironments of the two diseases. Conclusion The common pathogenesis of AD and DM was revealed in our research. These common pathways and hub genes directions for further exploration of the pathogenesis or treatment of these two diseases.
Collapse
Affiliation(s)
- Xian-Wen Ye
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China.,Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Meng-Nan Liu
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xuan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shui-Qing Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun-Shuai Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-Ying Bai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lin-Lin Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Xing Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Wen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wen-Juan Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shu-Yan Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Fang Xu
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China.,Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiang-Ri Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China.,Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Kulminski AM, Feng F, Loiko E, Nazarian A, Loika Y, Culminskaya I. Prevailing Antagonistic Risks in Pleiotropic Associations with Alzheimer's Disease and Diabetes. J Alzheimers Dis 2023; 94:1121-1132. [PMID: 37355909 PMCID: PMC10666173 DOI: 10.3233/jad-230397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2023]
Abstract
BACKGROUND The lack of efficient preventive interventions against Alzheimer's disease (AD) calls for identifying efficient modifiable risk factors for AD. As diabetes shares many pathological processes with AD, including accumulation of amyloid plaques and neurofibrillary tangles, insulin resistance, and impaired glucose metabolism, diabetes is thought to be a potentially modifiable risk factor for AD. Mounting evidence suggests that links between AD and diabetes may be more complex than previously believed. OBJECTIVE To examine the pleiotropic architecture of AD and diabetes mellitus (DM). METHODS Univariate and pleiotropic analyses were performed following the discovery-replication strategy using individual-level data from 10 large-scale studies. RESULTS We report a potentially novel pleiotropic NOTCH2 gene, with a minor allele of rs5025718 associated with increased risks of both AD and DM. We confirm previously identified antagonistic associations of the same variants with the risks of AD and DM in the HLA and APOE gene clusters. We show multiple antagonistic associations of the same variants with AD and DM in the HLA cluster, which were not explained by the lead SNP in this cluster. Although the ɛ2 and ɛ4 alleles played a major role in the antagonistic associations with AD and DM in the APOE cluster, we identified non-overlapping SNPs in this cluster, which were adversely and beneficially associated with AD and DM independently of the ɛ2 and ɛ4 alleles. CONCLUSION This study emphasizes differences and similarities in the heterogeneous genetic architectures of AD and DM, which may differentiate the pathogenic mechanisms of these diseases.
Collapse
Affiliation(s)
- Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Fan Feng
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Elena Loiko
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| |
Collapse
|
16
|
Sullivan M, Deng HW, Greenbaum J. Identification of genetic loci shared between Alzheimer's disease and hypertension. Mol Genet Genomics 2022; 297:1661-1670. [PMID: 36069947 DOI: 10.1007/s00438-022-01949-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/27/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) and high blood pressure (BP) are prevalent age-related diseases with significant unexplained heritability. A thorough analysis of genetic pleiotropy between AD and BP will lay a foundation for the study of the associated molecular mechanisms, leading to a better understanding of the development of each phenotype. We used the conditional false discovery rate (cFDR) method to identify novel genetic loci associated with both AD and BP. The cFDR approach improves the effective sample size for association testing by combining GWAS summary statistics for correlated phenotypes. We identified 50 pleiotropic SNPs for AD and BP, 7 of which are novel and have not previously been reported to be associated with either AD or BP. The novel SNPs located at STK3 are particularly noteworthy, as this gene may influence AD risk via the Hippo signaling network, which regulates cell death. Bayesian colocalization analysis demonstrated that although AD and BP are associated, they do not appear to share the same causal variants. We further performed two sample Mendelian randomization analysis, but could not detect a causal effect of BP on AD. Despite the inability to establish a causal link between AD and BP, our findings report some potential novel pleiotropic loci that may influence disease susceptibility. In summary, we identified 7 SNPs that annotate to 4 novel genes which have not previously been reported to be associated with AD nor with BP and discuss the possible role of one of these genes, STK3 in the Hippo signaling network.
Collapse
Affiliation(s)
- Megan Sullivan
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Jonathan Greenbaum
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
17
|
Yang J, Wang Z, Fu Y, Xu J, Zhang Y, Qin W, Zhang Q. Prediction value of the genetic risk of type 2 diabetes on the amnestic mild cognitive impairment conversion to Alzheimer’s disease. Front Aging Neurosci 2022; 14:964463. [PMID: 36185474 PMCID: PMC9521369 DOI: 10.3389/fnagi.2022.964463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
Amnestic mild cognitive impairment (aMCI) and Type 2 diabetes mellitus (T2DM) are both important risk factors for Alzheimer’s disease (AD). We aimed to investigate whether a T2DM-specific polygenic risk score (PRSsT2DM) can predict the conversion of aMCI to AD and further explore the underlying neurological mechanism. All aMCI patients were from the Alzheimer’s disease Neuroimaging Initiative (ADNI) database and were divided into conversion (aMCI-C, n = 164) and stable (aMCI-S, n = 222) groups. PRSsT2DM was calculated by PRSice-2 software to explore the predictive efficacy of the aMCI conversion to AD. We found that PRSsT2DM could independently predict the aMCI conversion to AD after removing the common variants of these two diseases. PRSsT2DM was significantly negatively correlated with gray matter volume (GMV) of the right superior frontal gyrus in the aMCI-C group. In all aMCI patients, PRSsT2DM was significantly negatively correlated with the cortical volume of the right superior occipital gyrus. The cortical volume of the right superior occipital gyrus could significantly mediate the association between PRSsT2DM and aMCI conversion. Gene-based analysis showed that T2DM-specific genes are highly expressed in cortical neurons and involved in ion and protein binding, neural development and generation, cell junction and projection, and PI3K-Akt and MAPK signaling pathway, which might increase the aMCI conversion by affecting the Tau phosphorylation and amyloid-beta (Aβ) accumulation. Therefore, the PRSsT2DM could be used as a measure to predict the conversion of aMCI to AD.
Collapse
|
18
|
Manzali SB, Yu E, Ravona-Springer R, Livny A, Golan S, Ouyang Y, Lesman-Segev O, Liu L, Ganmore I, Alkelai A, Gan-Or Z, Lin HM, Heymann A, Schnaider Beeri M, Greenbaum L. Alzheimer’s Disease Polygenic Risk Score Is Not Associated With Cognitive Decline Among Older Adults With Type 2 Diabetes. Front Aging Neurosci 2022; 14:853695. [PMID: 36110429 PMCID: PMC9468264 DOI: 10.3389/fnagi.2022.853695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesMultiple risk loci for late-onset Alzheimer’s disease (LOAD) have been identified. Type 2 diabetes (T2D) is a risk factor for cognitive decline, dementia and Alzheimer’s disease (AD). We investigated the association of polygenic risk score (PRS) for LOAD with overall cognitive functioning and longitudinal decline, among older adults with T2D.MethodsThe study included 1046 Jewish participants from the Israel Diabetes and Cognitive Decline (IDCD) study, aged ≥ 65 years, diagnosed with T2D, and cognitively normal at baseline. The PRS included variants from 26 LOAD associated loci (at genome-wide significance level), and was calculated with and without APOE. Outcome measures, assessed in 18 months intervals, were global cognition and the specific domains of episodic memory, attention/working memory, executive functions, and language/semantic categorization. Random coefficient models were used for analysis, adjusting for demographic variables, T2D-related characteristics, and cardiovascular factors. Additionally, in a subsample of 202 individuals, we analyzed the association of PRS with the volumes of total gray matter, frontal lobe, hippocampus, amygdala, and white matter hyperintensities. Last, the association of PRS with amyloid beta (Aβ) burden was examined in 44 participants who underwent an 18F-flutemetamol PET scan.ResultsThe PRS was not significantly associated with overall functioning or decline in global cognition or any of the specific cognitive domains. Similarly, following correction for multiple testing, there was no association with Aβ burden and other brain imaging phenotypes.ConclusionOur results suggest that the cumulative effect of LOAD susceptibility loci is not associated with a greater rate of cognitive decline in older adults with T2D, and other pathways may underlie this link.
Collapse
Affiliation(s)
- Sigalit B. Manzali
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Israel
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Ramit Ravona-Springer
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Memory Clinic, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abigail Livny
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Israel
| | - Sapir Golan
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuxia Ouyang
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Orit Lesman-Segev
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Israel
| | - Lang Liu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Ithamar Ganmore
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Memory Clinic, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anna Alkelai
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, United States
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Hung-Mo Lin
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anthony Heymann
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Maccabi Healthcare Services, Tel Aviv, Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lior Greenbaum
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel
- *Correspondence: Lior Greenbaum,
| |
Collapse
|
19
|
Exploring Lead loci shared between schizophrenia and Cardiometabolic traits. BMC Genomics 2022; 23:617. [PMID: 36008755 PMCID: PMC9414090 DOI: 10.1186/s12864-022-08766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 07/13/2022] [Indexed: 11/10/2022] Open
Abstract
Individuals with schizophrenia (SCZ) have, on average, a 10- to 20-year shorter expected life span than the rest of the population, primarily due to cardiovascular disease comorbidity. Genome-wide association studies (GWAS) have previously been used to separately identify common variants in SCZ and cardiometabolic traits. However, genetic variants jointly influencing both traits remain to be fully characterised. To assess overlaps (if any) between the genetic architecture of SCZ and cardiometabolic traits, we used conditional false discovery rate (FDR) and local genetic correlation statistical framework analyses. A conjunctional FDR was used to identify shared genetic traits between SCZ and cardiometabolic risk factors. We identified 144 genetic variants which were shared between SCZ and body mass index (BMI), and 15 variants shared between SCZ and triglycerides (TG). Furthermore, we discovered four novel single nucleotide polymorphisms (SNPs) (rs3865350, rs9860913, rs13307 and rs9614186) and four proximate genes (DERL2, SNX4, LY75 and EFCAB6) which were shared by SCZ and BMI. We observed that the novel genetic variant rs13307 and the most proximate gene LY75 exerted potential effects on SCZ and BMI comorbidity. Also, we observed a mixture of concordant and opposite direction associations with shared genetic variants. We demonstrated a moderate to high genetic overlap between SCZ and cardiometabolic traits associated with a pattern of bidirectional associations. Our data suggested a complex interplay between metabolism-related gene pathways in SCZ pathophysiology.
Collapse
|
20
|
Cheng Y, Li Y, Liang X, Wang P, Fa W, Liu C, Wang Y, Liu K, Wang N, Du Y. Genetic Effects of NDUFAF6 rs6982393 and APOE on Alzheimer’s Disease in Chinese Rural Elderly: A Cross-Sectional Population-Based Study. Clin Interv Aging 2022; 17:185-194. [PMID: 35237031 PMCID: PMC8884707 DOI: 10.2147/cia.s345784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/31/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate the associations of genotypes of NDUFAF6 rs6982393 and APOE and their combined genotypes with the risk of Alzheimer’s disease (AD) and mild cognitive impairment (MCI) in Chinese rural elderly. Methods This cross-sectional population-based study included 5096 older adults (age ≥60 years, 57.1% female). Genotypes of NDUFAF6 rs6982393 and APOE were detected using the multiple-polymerase chain reaction amplification. We diagnosed AD following the criteria of Diagnostic and Statistical Manual of Mental Disorders, the fourth edition and diagnosed MCI following the Petersen’s criteria MCI. Data were analyzed using the logistic regression model. Results The overall prevalence of AD and MCI was 3.57% (95% confidence interval [CI]: 0.040, 0.053) and 22.65% (95% CI: 0.223, 0.247), separately. The TT versus CC/CT genotype of NDUFAF6 rs6982393 was related to a higher risk of AD with the multi-adjusted odds ratio (95% CI) being 1.61 (1.02, 2.54) in the total sample, 3.36 (1.48, 7.60) in those aged 60–69, and 1.24 (0.71, 2.17) in those aged 70 years and above. The interaction between genotype of NDUFAF6 rs6982393 with age groups (60–69 versus ≥70 years) was significant on the risk of AD. The presence of APOE ε4 was not significantly associated with the risk of AD. Carrying both NDUFAF6 TT and APOE ε4 was related to a higher risk of AD with the multi-adjusted odds ratio (95% CI) being 2.69 (1.10, 2.56). In addition, there was no significant association between the above genotypes and MCI. Conclusion In Chinese rural elderly, the TT versus CT/CC genotype of NDUFAF6 rs6982393 was associated with an increased likelihood of AD; such an association only existed among young-old adults. Carrying both NDUFAF6 rs6982393-TT and APOE ε4 was related to a higher risk of AD. This finding highlights the importance of considering age and combined genotype in studying the genetic profiles of AD.
Collapse
Affiliation(s)
- Yingzhe Cheng
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Yuanjing Li
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Xiaoyan Liang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People’s Republic of China
| | - Pin Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Wenxin Fa
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Cuicui Liu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People’s Republic of China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People’s Republic of China
| | - Keke Liu
- Shandong Academy of Clinical Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Nan Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People’s Republic of China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, People’s Republic of China
- Correspondence: Yifeng Du, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, Shandong, 250021, People’s Republic of China, Tel/Fax +86-531-68776354, Email
| |
Collapse
|
21
|
Du H, Meng X, Yao Y, Xu J. The mechanism and efficacy of GLP-1 receptor agonists in the treatment of Alzheimer's disease. Front Endocrinol (Lausanne) 2022; 13:1033479. [PMID: 36465634 PMCID: PMC9714676 DOI: 10.3389/fendo.2022.1033479] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022] Open
Abstract
Since type 2 diabetes mellitus (T2DM) is a risk factor for Alzheimer's disease (AD) and both have the same pathogenesis (e.g., insulin resistance), drugs used to treat T2DM have been gradually found to reduce the progression of AD in AD models. Of these drugs, glucagon-like peptide 1 receptor (GLP-1R) agonists are more effective and have fewer side effects. GLP-1R agonists have reducing neuroinflammation and oxidative stress, neurotrophic effects, decreasing Aβ deposition and tau hyperphosphorylation in AD models, which may be a potential drug for the treatment of AD. However, this needs to be verified by further clinical trials. This study aims to summarize the current information on the mechanisms and effects of GLP-1R agonists in AD.
Collapse
Affiliation(s)
- Haiyang Du
- Division of Orthopedics, Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Meng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Yu Yao
- Division of Orthopedics, Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Xu
- Division of Orthopedics, Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jun Xu,
| |
Collapse
|
22
|
Dinh E, Rival T, Carrier A, Asfogo N, Corti O, Melon C, Salin P, Lortet S, Kerkerian-Le Goff L. TP53INP1 exerts neuroprotection under ageing and Parkinson's disease-related stress condition. Cell Death Dis 2021; 12:460. [PMID: 33966044 PMCID: PMC8106680 DOI: 10.1038/s41419-021-03742-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
TP53INP1 is a stress-induced protein, which acts as a dual positive regulator of transcription and of autophagy and whose deficiency has been linked with cancer and metabolic syndrome. Here, we addressed the unexplored role of TP53INP1 and of its Drosophila homolog dDOR in the maintenance of neuronal homeostasis under chronic stress, focusing on dopamine (DA) neurons under normal ageing- and Parkinson’s disease (PD)-related context. Trp53inp1−/− mice displayed additional loss of DA neurons in the substantia nigra compared to wild-type (WT) mice, both with ageing and in a PD model based on targeted overexpression of α-synuclein. Nigral Trp53inp1 expression of WT mice was not significantly modified with ageing but was markedly increased in the PD model. Trp53inp2 expression showed similar evolution and did not differ between WT and Trp53inp1−/− mice. In Drosophila, pan-neuronal dDOR overexpression improved survival under paraquat exposure and mitigated the progressive locomotor decline and the loss of DA neurons caused by the human α-synuclein A30P variant. dDOR overexpression in DA neurons also rescued the locomotor deficit in flies with RNAi-induced downregulation of dPINK1 or dParkin. Live imaging, confocal and electron microscopy in fat bodies, neurons, and indirect flight muscles showed that dDOR acts as a positive regulator of basal autophagy and mitophagy independently of the PINK1-mediated pathway. Analyses in a mammalian cell model confirmed that modulating TP53INP1 levels does not impact mitochondrial stress-induced PINK1/Parkin-dependent mitophagy. These data provide the first evidence for a neuroprotective role of TP53INP1/dDOR and highlight its involvement in the regulation of autophagy and mitophagy in neurons.
Collapse
Affiliation(s)
- Emilie Dinh
- Aix Marseille University, CNRS, IBDM, NeuroMarseille, Marseille, France
| | - Thomas Rival
- Aix Marseille University, CNRS, IBDM, NeuroMarseille, Marseille, France
| | - Alice Carrier
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Noemi Asfogo
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Olga Corti
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christophe Melon
- Aix Marseille University, CNRS, IBDM, NeuroMarseille, Marseille, France
| | - Pascal Salin
- Aix Marseille University, CNRS, IBDM, NeuroMarseille, Marseille, France
| | - Sylviane Lortet
- Aix Marseille University, CNRS, IBDM, NeuroMarseille, Marseille, France
| | | |
Collapse
|
23
|
Vogrinc D, Goričar K, Dolžan V. Genetic Variability in Molecular Pathways Implicated in Alzheimer's Disease: A Comprehensive Review. Front Aging Neurosci 2021; 13:646901. [PMID: 33815092 PMCID: PMC8012500 DOI: 10.3389/fnagi.2021.646901] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease, affecting a significant part of the population. The majority of AD cases occur in the elderly with a typical age of onset of the disease above 65 years. AD presents a major burden for the healthcare system and since population is rapidly aging, the burden of the disease will increase in the future. However, no effective drug treatment for a full-blown disease has been developed to date. The genetic background of AD is extensively studied; numerous genome-wide association studies (GWAS) identified significant genes associated with increased risk of AD development. This review summarizes more than 100 risk loci. Many of them may serve as biomarkers of AD progression, even in the preclinical stage of the disease. Furthermore, we used GWAS data to identify key pathways of AD pathogenesis: cellular processes, metabolic processes, biological regulation, localization, transport, regulation of cellular processes, and neurological system processes. Gene clustering into molecular pathways can provide background for identification of novel molecular targets and may support the development of tailored and personalized treatment of AD.
Collapse
Affiliation(s)
| | | | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
24
|
Ho WM, Wu YY, Chen YC. Genetic Variants behind Cardiovascular Diseases and Dementia. Genes (Basel) 2020; 11:genes11121514. [PMID: 33352859 PMCID: PMC7766236 DOI: 10.3390/genes11121514] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVDs) and dementia are the leading causes of disability and mortality. Genetic connections between cardiovascular risk factors and dementia have not been elucidated. We conducted a scoping review and pathway analysis to reveal the genetic associations underlying both CVDs and dementia. In the PubMed database, literature was searched using keywords associated with diabetes mellitus, hypertension, dyslipidemia, white matter hyperintensities, cerebral microbleeds, and covert infarctions. Gene lists were extracted from these publications to identify shared genes and pathways for each group. This included high penetrance genes and single nucleotide polymorphisms (SNPs) identified through genome wide association studies. Most risk SNPs to both diabetes and dementia participate in the phospholipase C enzyme system and the downstream nositol 1,4,5-trisphosphate and diacylglycerol activities. Interestingly, AP-2 (TFAP2) transcription factor family and metabolism of vitamins and cofactors were associated with genetic variants that were shared by white matter hyperintensities and dementia, and by microbleeds and dementia. Variants shared by covert infarctions and dementia were related to VEGF ligand-receptor interactions and anti-inflammatory cytokine pathways. Our review sheds light on future investigations into the causative relationships behind CVDs and dementia, and can be a paradigm of the identification of dementia treatments.
Collapse
Affiliation(s)
- Wei-Min Ho
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan; (W.-M.H.); (Y.-Y.W.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yah-Yuan Wu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan; (W.-M.H.); (Y.-Y.W.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Chun Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan; (W.-M.H.); (Y.-Y.W.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8433)
| |
Collapse
|
25
|
El Bitar F, Al Sudairy N, Qadi N, Al Rajeh S, Alghamdi F, Al Amari H, Al Dawsari G, Alsubaie S, Al Sudairi M, Abdulaziz S, Al Tassan N. A Comprehensive Analysis of Unique and Recurrent Copy Number Variations in Alzheimer's Disease and its Related Disorders. Curr Alzheimer Res 2020; 17:926-938. [PMID: 33256577 DOI: 10.2174/1567205017666201130111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/20/2020] [Accepted: 10/29/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Copy number variations (CNVs) play an important role in the genetic etiology of various neurological disorders, including Alzheimer's disease (AD). Type 2 diabetes mellitus (T2DM) and major depressive disorder (MDD) were shown to have share mechanisms and signaling pathways with AD. OBJECTIVE We aimed to assess CNVs regions that may harbor genes contributing to AD, T2DM, and MDD in 67 Saudi familial and sporadic AD patients, with no alterations in the known genes of AD and genotyped previously for APOE. METHODS DNA was analyzed using the CytoScan-HD array. Two layers of filtering criteria were applied. All the identified CNVs were checked in the Database of Genomic Variants (DGV). RESULTS A total of 1086 CNVs (565 gains and 521 losses) were identified in our study. We found 73 CNVs harboring genes that may be associated with AD, T2DM or MDD. Nineteen CNVs were novel. Most importantly, 42 CNVs were unique in our studied cohort existing only in one patient. Two large gains on chromosomes 1 and 13 harbored genes implicated in the studied disorders. We identified CNVs in genes that encode proteins involved in the metabolism of amyloid-β peptide (AGRN, APBA2, CR1, CR2, IGF2R, KIAA0125, MBP, RER1, RTN4R, VDR and WISPI) or Tau proteins (CACNAIC, CELF2, DUSP22, HTRA1 and SLC2A14). CONCLUSION The present work provided information on the presence of CNVs related to AD, T2DM, and MDD in Saudi Alzheimer's patients.
Collapse
Affiliation(s)
- Fadia El Bitar
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nourah Al Sudairy
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Najeeb Qadi
- Department of Neurosciences, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Fatimah Alghamdi
- Institute of Biology and Environmental Research, National Center for Biotechnology, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Hala Al Amari
- Institute of Biology and Environmental Research, National Center for Biotechnology, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Ghadeer Al Dawsari
- Institute of Biology and Environmental Research, National Center for Genomics Technology, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Sahar Alsubaie
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mishael Al Sudairi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Sara Abdulaziz
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nada Al Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Suresh J, Khor IW, Kaur P, Heng HL, Torta F, Dawe GS, Tai ES, Tolwinski NS. Shared signaling pathways in Alzheimer’s and metabolic disease may point to new treatment approaches. FEBS J 2020; 288:3855-3873. [DOI: 10.1111/febs.15540] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022]
Affiliation(s)
| | - Ing Wei Khor
- Department of Medicine Yong Loo Lin School of MedicineNational University of Singapore
| | - Prameet Kaur
- Science Division Yale‐ NUS College Singapore Singapore
| | - Hui Li Heng
- Department of Pharmacology Yong Loo Lin School of Medicine National University of Singapore, and Neurobiology Programme
- Life Sciences Institute National University of Singapore Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator Department of Biochemistry Yong Loo Lin School of MedicineNational University of Singapore Singapore
| | - Gavin S. Dawe
- Department of Pharmacology Yong Loo Lin School of Medicine National University of Singapore, and Neurobiology Programme
- Life Sciences Institute National University of Singapore Singapore
| | - E Shyong Tai
- Department of Medicine Yong Loo Lin School of MedicineNational University of Singapore
- Division of Endocrinology National University HospitalNational University Health System
| | | |
Collapse
|
27
|
Söderbom G, Zeng BY. The NLRP3 inflammasome as a bridge between neuro-inflammation in metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:345-391. [PMID: 32739011 DOI: 10.1016/bs.irn.2020.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Evidence increasingly suggests that type 2 diabetes mellitus (T2DM) is a risk factor for neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases share many pathological processes, including oxidative stress, local inflammation/neuroinflammation and chronic, low-grade (systemic) inflammation, which are exacerbated by aging, a common risk factor for T2DM and NDDs. Here, we focus on the link between chronic inflammation driven by peripheral metabolic disease and how this may impact neurodegeneration in AD and PD. We review the relationship between these common pathological processes in AD and PD from the perspective of the "pro-inflammatory" signaling of the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat- (LRR)-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complex. Since the need for effective disease-modifying therapies in T2DM, AD and PD is significant, the relationship between these diseases is important as a positive clinical impact on one may benefit the others. We briefly consider how novel strategies may target neuro-inflammation and provide potential therapies for AD and PD.
Collapse
Affiliation(s)
| | - Bai-Yun Zeng
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
28
|
Lutz MW, Luo S, Williamson DE, Chiba-Falek O. Shared genetic etiology underlying late-onset Alzheimer's disease and posttraumatic stress syndrome. Alzheimers Dement 2020; 16:1280-1292. [PMID: 32588970 DOI: 10.1002/alz.12128] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Late-onset Alzheimer's disease (LOAD) manifests comorbid neuropsychiatric symptoms and posttraumatic stress disorder (PTSD) is associated with an increased risk for dementia in late life, suggesting the two disorders may share genetic etiologies. METHODS We performed genetic pleiotropy analysis using LOAD and PTSD genome-wide association study (GWAS) datasets from white and African-American populations, followed by functional-genomic analyses. RESULTS We found an enrichment for LOAD across increasingly stringent levels of significance with the PTSD GWAS association (LOAD|PTSD) in the discovery and replication cohorts and a modest enrichment for the reverse conditional association (PTSD|LOAD). LOAD|PTSD association analysis identified and replicated the MS4A genes region. These genes showed similar expression pattern in brain regions affected in LOAD, and across-brain-tissue analysis identified a significant association for MS4A6A. The African-American samples showed moderate enrichment; however, no false discovery rate-significant associations. DISCUSSION We demonstrated common genetic signatures for LOAD and PTSD and suggested immune response as a common pathway for these diseases.
Collapse
Affiliation(s)
- Michael W Lutz
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | - Douglas E Williamson
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, USA.,Research Service, Durham VA Medical Center, Durham, North Carolina, USA.,Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA.,Center for Genomic and Computational Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
29
|
Caputo V, Termine A, Strafella C, Giardina E, Cascella R. Shared (epi)genomic background connecting neurodegenerative diseases and type 2 diabetes. World J Diabetes 2020; 11:155-164. [PMID: 32477452 PMCID: PMC7243483 DOI: 10.4239/wjd.v11.i5.155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/10/2020] [Accepted: 03/22/2020] [Indexed: 02/05/2023] Open
Abstract
The progressive aging of populations has resulted in an increased prevalence of chronic pathologies, especially of metabolic, neurodegenerative and movement disorders. In particular, type 2 diabetes (T2D), Alzheimer’s disease (AD) and Parkinson’s disease (PD) are among the most prevalent age-related, multifactorial pathologies that deserve particular attention, given their dramatic impact on patient quality of life, their economic and social burden as well the etiopathogenetic mechanisms, which may overlap in some cases. Indeed, the existence of common triggering factors reflects the contribution of mutual genetic, epigenetic and environmental features in the etiopathogenetic mechanisms underlying T2D and AD/PD. On this subject, this review will summarize the shared (epi)genomic features that characterize these complex pathologies. In particular, genetic variants and gene expression profiles associated with T2D and AD/PD will be discussed as possible contributors to determine the susceptibility and progression to these disorders. Moreover, potential shared epigenetic modifications and factors among T2D, AD and PD will also be illustrated. Overall, this review shows that findings from genomic studies still deserves further research to evaluate and identify genetic factors that directly contribute to the shared etiopathogenesis. Moreover, a common epigenetic background still needs to be investigated and characterized. The evidences discussed in this review underline the importance of integrating large-scale (epi)genomic data with additional molecular information and clinical and social background in order to finely dissect the complex etiopathogenic networks that build up the “disease interactome” characterizing T2D, AD and PD.
Collapse
Affiliation(s)
- Valerio Caputo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
| | - Andrea Termine
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
- Experimental and Behavioral Neurophysiology Laboratory, Santa Lucia Foundation, Rome 00142, Italy
| | - Claudia Strafella
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
| | - Emiliano Giardina
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation, Rome 00142, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
| | - Raffaella Cascella
- Department of Biomedicine and Prevention, Tor Vergata University, Rome 00133, Italy
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, Tirana 1000, Albania
| |
Collapse
|
30
|
Identification of novel functional CpG-SNPs associated with type 2 diabetes and coronary artery disease. Mol Genet Genomics 2020; 295:607-619. [PMID: 32162118 DOI: 10.1007/s00438-020-01651-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/03/2020] [Indexed: 02/08/2023]
Abstract
Genome-wide association studies (GWASs) have identified hundreds of single nucleotide polymorphisms (SNPs) associated with type 2 diabetes (T2D) and coronary artery disease (CAD), respectively. Nevertheless, these studies were generally performed for single-trait/disease and failed to assess the pleiotropic role of the identified variants. To identify novel functional loci and the pleiotropic relationship between CAD and T2D, the targeted cFDR analysis on CpG-SNPs was performed by integrating two independent large and multi-centered GWASs with summary statistics of T2D (26,676 cases and 132,532 controls) and CAD (60,801 cases and 123,504 controls). Applying the cFDR significance threshold of 0.05, we observed a pleiotropic enrichment between T2D and CAD by incorporating pleiotropic effects into a conditional analysis framework. We identified 79 novel CpG-SNPs for T2D, 61 novel CpG-SNPs for CAD, and 18 novel pleiotropic loci for both traits. Among these novel CpG-SNPs, 33 of them were annotated as methylation quantitative trait locus (meQTL) in whole blood, and ten of them showed expression QTL (eQTL), meQTL, and metabolic QTL (metaQTL) effects simultaneously. To the best of our knowledge, we performed the first targeted cFDR analysis on CpG-SNPs, and our findings provided novel insights into the shared biological mechanisms and overlapped genetic heritability between T2D and CAD.
Collapse
|
31
|
Shared genetic etiology underlying Alzheimer's disease and major depressive disorder. Transl Psychiatry 2020; 10:88. [PMID: 32152295 PMCID: PMC7062839 DOI: 10.1038/s41398-020-0769-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/14/2020] [Accepted: 02/25/2020] [Indexed: 01/22/2023] Open
Abstract
Patients with late-onset Alzheimer's disease (LOAD) frequently manifest comorbid neuropsychiatric symptoms with depression and anxiety being most frequent, and individuals with major depressive disorder (MDD) have an increased prevalence of LOAD. This suggests shared etiologies and intersecting pathways between LOAD and MDD. We performed pleiotropy analyses using LOAD and MDD GWAS data sets from the International Genomics of Alzheimer's Project (IGAP) and the Psychiatric Genomics Consortium (PGC), respectively. We found a moderate enrichment for SNPs associated with LOAD across increasingly stringent levels of significance with the MDD GWAS association (LOAD|MDD), of maximum four and eightfolds, including and excluding the APOE-region, respectively. Association analysis excluding the APOE-region identified numerous SNPs corresponding to 40 genes, 9 of which are known LOAD-risk loci primarily in chromosome 11 regions that contain the SPI1 gene and MS4A genes cluster, and others were novel pleiotropic risk-loci for LOAD conditional with MDD. The most significant associated SNPs on chromosome 11 overlapped with eQTLs found in whole-blood and monocytes, suggesting functional roles in gene regulation. The reverse conditional association analysis (MDD|LOAD) showed a moderate level, ~sevenfold, of polygenic overlap, however, no SNP showed significant association. Pathway analyses replicated previously reported LOAD biological pathways related to immune response and regulation of endocytosis. In conclusion, we provide insights into the overlapping genetic signatures underpinning the common phenotypic manifestations and inter-relationship between LOAD and MDD. This knowledge is crucial to the development of actionable targets for novel therapies to treat depression preceding dementia, in an effort to delay or ultimately prevent the onset of LOAD.
Collapse
|
32
|
Chen LC, Fan ZY, Wang HY, Wen DC, Zhang SY. Effect of polysaccharides from adlay seed on anti-diabetic and gut microbiota. Food Funct 2020; 10:4372-4380. [PMID: 31276140 DOI: 10.1039/c9fo00406h] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes is a chronic metabolic disease characterized by elevated blood glucose levels due to insulin resistance and β-cell dysfunction. This study aims to examine the effects of polysaccharides from adlay seeds (PAS) on hyperglycemia and gut microbiota in streptozocin (STZ)-induced diabetic mice. The administration of PAS in diabetic mice caused a significant decrease in the glucose level and serum levels of glycosylated hemoglobin (HbA1c). Similarly, PAS also showed decreased total cholesterol (TC) and triglyceride (TG) concentrations. Furthermore, a significant increase in the concentrations of glucagon-like peptide 1 (GLP-1) was observed. Unexpectedly, PAS reduced the concentrations of anti-amyloid beta (Aβ1-42) protein. Also, histopathological examination showed that PAS contributed to the reduction of STZ-lesioned pancreatic cells. Metformin treatment significantly reduced the diversity of the gut microbiota, while PAS treatment altered the diversity and composition of the microbiota. Collectively, our findings demonstrate that the hypoglycemic effects of PAS in type-2 diabetic mice (T2D) may be associated with the regulation of the intestinal microbiota and its metabolic pathways.
Collapse
Affiliation(s)
- Li-Chun Chen
- College of Food & Biology Engineering, Zhejiang Gongshang University, Hangzhou, Zhejiang 310035, China.
| | | | | | | | | |
Collapse
|
33
|
Chen LC, Zhang SY, Zi Y, Zhao HM, Wang HY, Zhang Y. Functional coix seed protein hydrolysates as a novel agent with potential hepatoprotective effect. Food Funct 2020; 11:9495-9502. [DOI: 10.1039/d0fo01658f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The aim of this study is to explore the hepatoprotective potential of coix seed protein hydrolysates (CPP) against alcohol-induced liver injury, and investigate the underlying mechanisms.
Collapse
Affiliation(s)
- Li-Chun Chen
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products
| | - Shi-Yu Zhang
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products
- Zhejiang University of Science and Technology
- Hangzhou
- China
| | - Yu Zi
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Hui-Min Zhao
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Hong-Yu Wang
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products
- Zhejiang University of Science and Technology
- Hangzhou
- China
| | - Yue Zhang
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| |
Collapse
|
34
|
Chen LC, Jiang BK, Zheng WH, Zhang SY, Li JJ, Fan ZY. Preparation, characterization and anti-diabetic activity of polysaccharides from adlay seed. Int J Biol Macromol 2019; 139:605-613. [PMID: 31381909 DOI: 10.1016/j.ijbiomac.2019.08.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/12/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
Coix (Coix lachryma-jobi L.), commonly known as adlay, is a traditional Chinese medicine for thousands of years. A new water-soluble polysaccharide with anti-diabetic activity was extracted and purified from the adlay seed (PAS). The structure and physicochemical properties of PAS were determined by Fourier transform infrared spectrometer (FT-IR) and scanning electron microscopy (SEM). Structural analysis indicated that PAS had a porous surface and relatively loose distribution. After intragastric administered PAS for 4 weeks, biochemical analysis demonstrated dose dependent anti-diabetic activity. These results showed that PAS decreased blood glucose and insulin levels. In addition, mice fed the PAS showed significantly reduced the plasma levels of amyloid β42 and glycated hemoglobin (HbA1c), while the expression of glucagon-like peptide-1 (GLP-1) was markedly increased. Our study introduced a new polysaccharide PAS with unique anti-diabetic activity, which can be used as a potential dietary supplement or functional food.
Collapse
Affiliation(s)
- Li-Chun Chen
- College of Food & Biology Engineering, Zhejiang Gongshang University, Hangzhou, Zhejiang 310035, China.
| | - Bo-Kai Jiang
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Wen-Hao Zheng
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Shi-Yu Zhang
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Jia-Jiang Li
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Zhong-Yang Fan
- Zhejiang Provincial Key Lab for Chem &Bio Processing Technology of Agricultural Products, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| |
Collapse
|
35
|
Molecular Connection Between Diabetes and Dementia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:103-131. [DOI: 10.1007/978-981-13-3540-2_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Alzheimer's disease in the omics era. Clin Biochem 2018; 59:9-16. [DOI: 10.1016/j.clinbiochem.2018.06.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022]
|
37
|
Leone S, Recinella L, Chiavaroli A, Orlando G, Ferrante C, Leporini L, Brunetti L, Menghini L. Phytotherapic use of theCrocus sativusL. (Saffron) and its potential applications: A brief overview. Phytother Res 2018; 32:2364-2375. [DOI: 10.1002/ptr.6181] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/03/2018] [Accepted: 07/22/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Sheila Leone
- Department of Pharmacy; G. d'Annunzio University; Chieti Italy
| | - Lucia Recinella
- Department of Pharmacy; G. d'Annunzio University; Chieti Italy
| | | | | | | | - Lidia Leporini
- Department of Pharmacy; G. d'Annunzio University; Chieti Italy
| | - Luigi Brunetti
- Department of Pharmacy; G. d'Annunzio University; Chieti Italy
| | - Luigi Menghini
- Department of Pharmacy; G. d'Annunzio University; Chieti Italy
| |
Collapse
|
38
|
Verma MK, Goel R, Krishnadas N, Nemmani KVS. Targeting glucose-dependent insulinotropic polypeptide receptor for neurodegenerative disorders. Expert Opin Ther Targets 2018; 22:615-628. [PMID: 29911915 DOI: 10.1080/14728222.2018.1487952] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Incretin hormones, glucose-dependent insulinotropic polypeptide (GIP), and glucagon-like peptide-1 (GLP-1) exert pleiotropic effects on endocrine pancreas and nervous system. Expression of GIP and GIP receptor (GIPR) in neurons, their roles in neurogenesis, synaptic plasticity, neurotransmission, and neuromodulation uniquely position GIPR for therapeutic applications in neurodegenerative disorders. GIP analogs acting as GIPR agonists attenuate neurobehavioral and neuropathological sequelae of neurodegenerative disorders in preclinical models, e.g. Alzheimer's disease (AD), Parkinson's disease (PD), and cerebrovascular disorders. Modulation of GIPR signaling offers an unprecedented approach for disease modification by arresting neuronal viability decline, enabling neuronal regeneration, and reducing neuroinflammation. Growth-promoting effects of GIP signaling and broad-based neuroprotection highlight the therapeutic potential of GIPR agonists. Areas covered: This review focuses on the role of GIPR-mediated signaling in the central nervous system in neurophysiological and neuropathological conditions. In context of neurodegeneration, the article summarizes potential of targeting GIPR signaling for neurodegenerative conditions such as AD, PD, traumatic brain injury, and cerebrovascular disorders. Expert opinion: GIPR represents a validated therapeutic target for neurodegenerative disorders. GIPR agonists impart symptomatic improvements, slowed neurodegeneration, and enhanced neuronal regenerative capacity in preclinical models. Modulation of GIPR signaling is potentially a viable therapeutic approach for disease modification in neurodegenerative disorders.
Collapse
Affiliation(s)
- Mahip K Verma
- a Department of Pharmacology, Novel Drug Discovery and Development , Lupin Limited , Pune , India
| | - Rajan Goel
- a Department of Pharmacology, Novel Drug Discovery and Development , Lupin Limited , Pune , India
| | - Nandakumar Krishnadas
- b Department of Pharmacology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE) , Manipal , India
| | - Kumar V S Nemmani
- a Department of Pharmacology, Novel Drug Discovery and Development , Lupin Limited , Pune , India
| |
Collapse
|
39
|
Alimoradian A, Ghasemi S, Zahiri M, Saeedi AH, Miladi H, Sadegh M. Investigation of the effect of Ginkgo biloba leaf extract on spatial memory impairment and hippocampal neuronal loss caused by diabetes induced by streptozotocin in rats. SCIENTIFIC JOURNAL OF KURDISTAN UNIVERSITY OF MEDICAL SCIENCES 2018. [DOI: 10.29252/sjku.23.2.114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
40
|
Menghini L, Leporini L, Vecchiotti G, Locatelli M, Carradori S, Ferrante C, Zengin G, Recinella L, Chiavaroli A, Leone S, Brunetti L, Orlando G. Crocus sativus L. stigmas and byproducts: Qualitative fingerprint, antioxidant potentials and enzyme inhibitory activities. Food Res Int 2018; 109:91-98. [PMID: 29803496 DOI: 10.1016/j.foodres.2018.04.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/05/2018] [Accepted: 04/14/2018] [Indexed: 02/08/2023]
Abstract
Saffron (Crocus sativus L.) has been previously reported to be active as a protective agent in multiple experimental models of oxidative stress, inflammation and cancer. These findings refer to the protective effects of stigmas, not byproducts such as tepals and anthers. In this context, the aims of the present work were to characterize the phytochemical profile of saffron stigmas (CST) and high quality byproducts (tepals + anthers - CTA) extracts. Additionally, we studied the antioxidant and chelating effects of CST and CTA extracts by preliminary in vitro assay. The antioxidant activity was further investigated through the evaluation of reactive oxygen species (ROS) levels and lactate dehydrogenase (LDH) activity on mouse myoblast (C2C12) and human colon cancer (HCT116) cell lines. Additionally, we evaluated CST and CTA extract treatment on cholinesterases, α-glucosidase and α-amylase activity, in vitro. Finally, we studied the effects of CST extract on malondialdehyde (MDA) level in rat colon specimens challenged with E. coli lipopolysaccharide (LPS). We observed that water CST extracts are rich in phenolic content, whereas for CTA the olive oil was the elective extraction solvent. As expected, water CST extracts were the most effective in reducing hydrogen peroxide-induced oxidative stress in both cell lines and in vitro assays. Furthermore, both CST and CTA water extracts reduced the LDH activity in HCT116 cells challenged with hydrogen peroxide and LPS-induced MDA levels in rat colon specimens. Concluding, the present findings showed protective effects exerted by CST and CTA extracts in in vitro and ex vivo models of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Luigi Menghini
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Lidia Leporini
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Giulia Vecchiotti
- Dipartimento di Medicina Clinica, Sanità Pubblicà, Scienze della Vita e dell'Ambiente, Università dell'Aquila, L'Aquila, Italy
| | - Marcello Locatelli
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Simone Carradori
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Claudio Ferrante
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.
| | - Gokhan Zengin
- Selcuk University, Science Faculty, Department of Biology, Konya, Turkey
| | - Lucia Recinella
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Annalisa Chiavaroli
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Sheila Leone
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Luigi Brunetti
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Giustino Orlando
- Department of Pharmacy, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
41
|
Lim PH, Wert SL, Tunc-Ozcan E, Marr R, Ferreira A, Redei EE. Premature hippocampus-dependent memory decline in middle-aged females of a genetic rat model of depression. Behav Brain Res 2018; 353:242-249. [PMID: 29490235 DOI: 10.1016/j.bbr.2018.02.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/01/2018] [Accepted: 02/21/2018] [Indexed: 01/08/2023]
Abstract
Aging and major depressive disorder are risk factors for dementia, including Alzheimer's Disease (AD), but the mechanism(s) linking depression and dementia are not known. Both AD and depression show greater prevalence in women. We began to investigate this connection using females of the genetic model of depression, the inbred Wistar Kyoto More Immobile (WMI) rat. These rats consistently display depression-like behavior compared to the genetically close control, the Wistar Kyoto Less Immobile (WLI) strain. Hippocampus-dependent contextual fear memory did not differ between young WLI and WMI females, but, by middle-age, female WMIs showed memory deficits compared to same age WLIs. This deficit, measured as duration of freezing in the fear provoking-context was not related to activity differences between the strains prior to fear conditioning. Hippocampal expression of AD-related genes, such as amyloid precursor protein, amyloid beta 42, beta secretase, synucleins, total and dephosphorylated tau, and synaptophysin, did not differ between WLIs and WMIs in either age group. However, hippocampal transcript levels of catalase (Cat) and hippocampal and frontal cortex expression of insulin-like growth factor 2 (Igf2) and Igf2 receptor (Igf2r) paralleled fear memory differences between middle-aged WLIs and WMIs. This data suggests that chronic depression-like behavior that is present in this genetic model is a risk factor for early spatial memory decline in females. The molecular mechanisms of this early memory decline likely involve the interaction of aging processes with the genetic components responsible for the depression-like behavior in this model.
Collapse
Affiliation(s)
- Patrick H Lim
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Stephanie L Wert
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Elif Tunc-Ozcan
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Robert Marr
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States
| | - Adriana Ferreira
- Department of Cellular and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States.
| |
Collapse
|
42
|
Wilkins JM, Trushina E. Application of Metabolomics in Alzheimer's Disease. Front Neurol 2018; 8:719. [PMID: 29375465 PMCID: PMC5770363 DOI: 10.3389/fneur.2017.00719] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
Progress toward the development of efficacious therapies for Alzheimer’s disease (AD) is halted by a lack of understanding early underlying pathological mechanisms. Systems biology encompasses several techniques including genomics, epigenomics, transcriptomics, proteomics, and metabolomics. Metabolomics is the newest omics platform that offers great potential for the diagnosis and prognosis of neurodegenerative diseases as an individual’s metabolome reflects alterations in genetic, transcript, and protein profiles and influences from the environment. Advancements in the field of metabolomics have demonstrated the complexity of dynamic changes associated with AD progression underscoring challenges with the development of efficacious therapeutic interventions. Defining systems-level alterations in AD could provide insights into disease mechanisms, reveal sex-specific changes, advance the development of biomarker panels, and aid in monitoring therapeutic efficacy, which should advance individualized medicine. Since metabolic pathways are largely conserved between species, metabolomics could improve the translation of preclinical research conducted in animal models of AD into humans. A summary of recent developments in the application of metabolomics to advance the AD field is provided below.
Collapse
Affiliation(s)
- Jordan Maximillian Wilkins
- Mitochondrial Neurobiology and Therapeutics Laboratory, Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Eugenia Trushina
- Mitochondrial Neurobiology and Therapeutics Laboratory, Department of Neurology, Mayo Clinic, Rochester, MN, United States.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
43
|
Hu Y, Tan LJ, Chen XD, Liu Z, Min SS, Zeng Q, Shen H, Deng HW. Identification of Novel Potentially Pleiotropic Variants Associated With Osteoporosis and Obesity Using the cFDR Method. J Clin Endocrinol Metab 2018; 103:125-138. [PMID: 29145611 PMCID: PMC6061219 DOI: 10.1210/jc.2017-01531] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/12/2017] [Indexed: 01/10/2023]
Abstract
CONTEXT Genome-wide association studies (GWASs) have been successful in identifying loci associated with osteoporosis and obesity. However, the findings explain only a small fraction of the total genetic variance. OBJECTIVE The aim of this study was to identify novel pleiotropic genes important in osteoporosis and obesity. DESIGN AND SETTING A pleiotropic conditional false discovery rate method was applied to three independent GWAS summary statistics of femoral neck bone mineral density, body mass index, and waist-to-hip ratio. Next, differential expression analysis was performed for the potentially pleiotropic genes, and weighted genes coexpression network analysis (WGCNA) was conducted to identify functional connections between the suggested pleiotropic genes and known osteoporosis/obesity genes using transcriptomic expression data sets in osteoporosis/obesity-related cells. RESULTS We identified seven potentially pleiotropic loci-rs3759579 (MARK3), rs2178950 (TRPS1), rs1473 (PUM1), rs9825174 (XXYLT1), rs2047937 (ZNF423), rs17277372 (DNM3), and rs335170 (PRDM6)-associated with osteoporosis and obesity. Of these loci, the PUM1 gene was differentially expressed in osteoporosis-related cells (B lymphocytes) and obesity-related cells (adipocytes). WGCNA showed that PUM1 positively interacted with several known osteoporosis genes (AKAP11, JAG1, and SPTBN1). ZNF423 was the highly connected intramodular hub gene and interconnected with 21 known osteoporosis-related genes, including JAG1, EN1, and FAM3C. CONCLUSIONS Our study identified seven potentially pleiotropic genes associated with osteoporosis and obesity. The findings may provide new insights into a potential genetic determination and codetermination mechanism of osteoporosis and obesity.
Collapse
Affiliation(s)
- Yuan Hu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Li-Jun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiang-Ding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Zhen Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Shi-Shi Min
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Qin Zeng
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Hui Shen
- Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Hong-Wen Deng
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|